1
|
Liao J, Gu Q, Liu Z, Wang H, Yang X, Yan R, Zhang X, Song S, Wen L, Wang Y. Edge advances in nanodrug therapies for osteoarthritis treatment. Front Pharmacol 2024; 15:1402825. [PMID: 39539625 PMCID: PMC11559267 DOI: 10.3389/fphar.2024.1402825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
As global population and lifestyles change, osteoarthritis (OA) is becoming a major healthcare challenge world. OA, a chronic condition characterized by inflammatory and degeneration, often present with joint pain and can lead to irreversible disability. While there is currently no cure for OA, it is commonly managed using nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, and glucosamine. Although these treatments can alleviate symptoms, it is difficult to effectively deliver and sustain therapeutic agents within joints. The emergence of nanotechnology, particularly in form of smart nanomedicine, has introduced innovative therapeutic approaches for OA treatment. Nanotherapeutic strategies offer promising advantages, including more precise targeting of affected areas, prolonged therapeutic effects, enhanced bioavailability, and reduced systemic toxicity compared to traditional treatments. While nanoparticles show potential as a viable delivery system for OA therapies based on encouraging lab-based and clinical trials results, there remails a considerable gap between current research and clinical application. This review highlights recent advances in nanotherapy for OA and explore future pathways to refine and optimize OA treatments strategies.
Collapse
Affiliation(s)
- Jinfeng Liao
- Department of Dermatology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Qingjia Gu
- Department of ENT, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Zheng Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Xian Yang
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongkai Yan
- Department of Radiology, Ohio state university, Columbus, OH, United States
| | - Xiaofeng Zhang
- Greenwich Hospital, Yale New Haven Health, Greenwich, CT, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Lebin Wen
- Department of Thyroid, Sichuan Second Hospital of TCM, Chengdu, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
2
|
Sheng JY, Meng ZF, Li Q, Yang YS. Recent advances in promising drugs for primary prevention of gastroesophageal variceal bleeding with cirrhotic portal hypertension. Hepatobiliary Pancreat Dis Int 2024; 23:4-13. [PMID: 37580228 DOI: 10.1016/j.hbpd.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Gastroesophageal variceal bleeding is one of the most severe complications of patients with cirrhosis. Although primary prevention drugs, including non-selective β-blockers, have effectively reduced the incidence of bleeding, their efficacy is limited due to side effects and related contraindications. With recent advances in precision medicine, precise drug treatment provides better treatment efficacy. DATA SOURCES Literature search was conducted in PubMed, MEDLINE and Web of Science for relevant articles published up to May 2022. Information on clinical trials was obtained from https://clinicaltrials.gov/ and http://www.chictr.org.cn/. RESULTS The in-depth understanding of the pathogenesis and advances of portal hypertension has enabled the discovery of multiple molecular targets for promising drugs. According to the site of action, these drugs could be classified into four classes: intrahepatic, extrahepatic, both intrahepatic and extrahepatic targets and others. All these classes of drugs offer advantages over traditional treatments in prevention of gastroesophageal variceal bleeding in patients with cirrhotic portal hypertension. CONCLUSIONS This review classified and summarized the promising drugs, which prevent gastroesophageal variceal bleeding by targeting specific markers of pathogenesis of portal hypertension, demonstrating the significance of using the precision medicine strategy to discover and develop promising drugs for the primary prevention of gastroesophageal variceal bleeding in patients with cirrhotic portal hypertension.
Collapse
Affiliation(s)
- Ji-Yao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China; Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, the Second Hospital of Jilin University, Changchun 130041, China
| | - Zi-Fan Meng
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China; Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, the Second Hospital of Jilin University, Changchun 130041, China
| | - Qiao Li
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Yong-Sheng Yang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China; Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, the Second Hospital of Jilin University, Changchun 130041, China.
| |
Collapse
|
3
|
Shihmani B, Rassouli A, Mehrzad J, Shokrpoor S. The anti-inflammatory effects of minocycline on lipopolysaccharide-induced paw oedema in rats: a histopathological and molecular study. Inflammopharmacology 2023:10.1007/s10787-023-01236-7. [PMID: 37119392 DOI: 10.1007/s10787-023-01236-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/10/2023] [Indexed: 05/01/2023]
Abstract
Minocycline is a semi-synthetic antimicrobial agent with claimed anti-inflammatory properties reported from different experimental models. This study was aimed to evaluate the anti-inflammatory effects of minocycline, compared to the actions of two common anti-inflammatory agents, on lipopolysaccharide (LPS)-induced paw oedema through some clinical, histopathological, haematological and molecular analyses. Forty-eight rats were divided into eight groups (n = 6). In control group (Ctrl), each animal was injected with normal saline into its sub-plantar region of hind paw. In groups 2-7, hind paw oedema was induced by injection of LPS. One hour before injections, groups 1 (Ctrl) and 2 (LPS) were treated orally with distilled water, 3 and 4 with methylprednisolone (Pred) and meloxicam (Melo) and 5-7 with minocycline in doses of 50, 150 and 450 mg/kg (M50, M150 and M450, respectively). The 8th group (MC) was given minocycline (150 mg/kg) orally and normal saline was injected into sub-plantar region. Paw swelling and body temperature were assessed at 0, 2, 4, 6 and 24 h post-injections. At 24 h, samples of blood and liver, kidney, spleen and hind paw tissues were taken for haematological and histopathological examinations. Some samples of the paw were also obtained for molecular analysis of some inflammatory-related cytokines at mRNA level. Paw swelling and body temperature increased in all LPS-injected groups 2 h post-injection. In LPS group, they remained significantly increased up to 24 h; however, these parameters decreased to normal in Pred, Melo and all minocycline groups. The histological findings showed mild-to-moderate signs of inflammation in tissue samples of groups 2-6, but not in group M450. Additionally, gene expression of pro-inflammatory cytokines (IL-1β and IL-6) increased significantly in LPS group compared to other groups. In conclusion, this study supports the role of minocycline as an anti-inflammatory agent with effects comparable to those of meloxicam and methylprednisolone.
Collapse
Affiliation(s)
- Basim Shihmani
- Pharmacology Division, Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 1419963114, Iran
| | - Ali Rassouli
- Pharmacology Division, Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 1419963114, Iran.
| | - Jalil Mehrzad
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Sara Shokrpoor
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
4
|
Cui L, Guo J, Wang Z, Zhang J, Li W, Dong J, Liu K, Guo L, Li J, Wang H, Li J. Meloxicam inhibited oxidative stress and inflammatory response of LPS-stimulated bovine endometrial epithelial cells through Nrf2 and NF-κB pathways. Int Immunopharmacol 2023; 116:109822. [PMID: 36750013 DOI: 10.1016/j.intimp.2023.109822] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/16/2023] [Accepted: 01/28/2023] [Indexed: 02/07/2023]
Abstract
Meloxicam is a selective cyclooxygenase-2 inhibitor and has been widely used in combination with antibiotics to alleviate uterine inflammation and provide analgesia in postpartum cows. Studies have shown that meloxicam has antioxidant and anti-inflammatory effects. However, the link between meloxicam and uterine inflammation and oxidative stress in dairy cows has not been studied. The purpose of this study was to research the effects of meloxicam (0.5 or 5 μM) on oxidative stress and inflammatory response of primary bovine endometrial epithelial cells (BEEC) stimulated by Escherichia coli lipopolysaccharide (1 μg/mL LPS). As a result, LPS stimulated the production of oxidative stress markers and the expression of inflammatory factors, accompanied by a decrease in the activity and the gene transcription of antioxidant enzymes. Co-treatment of meloxicam and LPS reduced the content of oxidative stress markers and the mRNA levels of the pro-inflammatory genes, and improved antioxidant enzyme activities and the corresponding gene expression as compared with the cells treated with LPS alone. Meloxicam attenuated the inhibitory effect of the Nrf2 pathway and the phosphorylation levels of p65 and IκBα caused by LPS. In conclusion, meloxicam alone had no effect on BEEC, but prevented oxidative stress and inflammatory response in LPS-stimulated BEEC.
Collapse
Affiliation(s)
- Luying Cui
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Jing Guo
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Zhihao Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Jiaqi Zhang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Wenjie Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Kangjun Liu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Long Guo
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Jun Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 220559, PR China; Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| |
Collapse
|
5
|
Miranda HF, Noriega V, Sierralta F, Sotomayor‐Zárate R, Prieto JC. Risperidone in analgesia induced by paracetamol and meloxicam in experimental pain. Fundam Clin Pharmacol 2022; 36:494-500. [DOI: 10.1111/fcp.12754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/10/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Hugo F. Miranda
- Neuroscience Department, Faculty of Medicine University of Chile Santiago 8380000 Chile
| | - Viviana Noriega
- Faculty of Medicine, German Cinic University of Development Santiago 8380000 Chile
- Cardiovascular Department University of Chile Clinical Hospital Santiago 8380000 Chile
| | - Fernando Sierralta
- Pharmacology Program, ICBM, Faculty of Medicine University of Chile Santiago 8380000 Chile
| | - Ramón Sotomayor‐Zárate
- Laboratory of Neurochemistry and Neuropharmacology, Faculty of Sciences University of Valparaíso Valparaíso 8380000 Chile
| | - Juan Carlos Prieto
- Cardiovascular Department University of Chile Clinical Hospital Santiago 8380000 Chile
- Pharmacology Program, ICBM, Faculty of Medicine University of Chile Santiago 8380000 Chile
| |
Collapse
|
6
|
Gad El-Hak HN, Mahmoud HS, Ahmed EA, Elnegris HM, Aldayel TS, Abdelrazek HMA, Soliman MTA, El-Menyawy MAI. Methanolic Phoenix dactylifera L. Extract Ameliorates Cisplatin-Induced Hepatic Injury in Male Rats. Nutrients 2022; 14:1025. [PMID: 35268000 PMCID: PMC8912432 DOI: 10.3390/nu14051025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/20/2022] Open
Abstract
This study investigated the ameliorative potential of methanolic date flesh extract (MDFE) against cisplatin-induced hepatic injury. Twenty male rats (weighing 180-200 g) were allocated into four groups: control; date flesh (DF) group (oral 600 mg/kg MDFE for 21 days); Cis group (7.5 mg/kg i.p. at day 16); and date flesh/cisplatin (DF/Cis) group (oral 600 mg/kg MDFE for 21 days and 7.5 mg/kg i.p. at day 16). Hepatic biochemical parameters in sera, and inflammatory and oxidant/antioxidant hepatic biomarkers were estimated. Hepatic histological changes and the immunohistochemistry of cyclooxygenase-2 (COX-2), nuclear factor kappa B (NF-κB), and alpha smooth muscle actin (α-SMA) were assessed. Pretreatment with MDFE decreased Cis-triggered liver biochemical parameters, oxidative stress, inflammatory biomarkers, and histological damage. Moreover, MDFE treatment reduced Cis-induced hepatic NF-κB, COX-2, and α-SMA protein expression. MDFE exerted a hepatoprotective effect when used concomitantly with Cis. Its effect was mediated via its antioxidant and anti-inflammatory ingredients.
Collapse
Affiliation(s)
- Heba Nageh Gad El-Hak
- Zoology Department, Faculty of Sciences, Suez Canal University, Ismailia 41522, Egypt;
| | - Hany Salah Mahmoud
- Center of Scientific Foundation for Experimental Studies and Research, Ismailia 41511, Egypt;
| | - Eman A. Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Heba M. Elnegris
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt;
- Department of Histology and Cell Biology, Faculty of Medicine, Badr University in Cairo, Cairo 11829, Egypt
| | - Tahany Saleh Aldayel
- Department of Physical Sport Sciences, College of Education, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Heba M. A. Abdelrazek
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Mohamed T. A. Soliman
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 67614, Saudi Arabia;
| | | |
Collapse
|
7
|
Tai Y, Zhao C, Zhang L, Tang S, Jia X, Tong H, Liu R, Tang C, Gao J. Celecoxib reduces hepatic vascular resistance in portal hypertension by amelioration of endothelial oxidative stress. J Cell Mol Med 2021; 25:10389-10402. [PMID: 34609050 PMCID: PMC8581330 DOI: 10.1111/jcmm.16968] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 02/05/2023] Open
Abstract
The balance between endothelial nitric oxide (NO) synthase (eNOS) activation and production of reactive oxygen species (ROS) is very important for NO homeostasis in liver sinusoidal endothelial cells (LSECs). Overexpression of cyclooxygenase‐2 (COX‐2), a major intravascular source of ROS production, has been observed in LSECs of cirrhotic liver. However, the links between low NO bioavailability and COX‐2 overexpression in LSECs are unknown. This study has confirmed the link between low NO bioavailability and COX‐2 overexpression by COX‐2‐dependent PGE2‐EP2‐ERK1/2‐NOX1/NOX4 signalling pathway in LSECs in vivo and in vitro. In addition, the regulation of COX‐2‐independent LKB1‐AMPK‐NRF2‐HO‐1 signalling pathway on NO homeostasis in LSECs was also elucidated. The combinative effects of celecoxib on diminishment of ROS via COX‐2‐dependent and COX‐2‐independent signalling pathways greatly decreased NO scavenging. As a result, LSECs capillarisation was reduced, and endothelial dysfunction was corrected. Furthermore, portal hypertension of cirrhotic liver was ameliorated with substantial decreasing hepatic vascular resistance and great increase of portal blood flow. With the advance understanding of the mechanisms of LSECs protection, celecoxib may serve as a potential therapeutic candidate for patients with cirrhotic portal hypertension.
Collapse
Affiliation(s)
- Yang Tai
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chong Zhao
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Linhao Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Shihang Tang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xintong Jia
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Tong
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Gao
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Cui L, Qu Y, Cai H, Wang H, Dong J, Li J, Qian C, Li J. Meloxicam Inhibited the Proliferation of LPS-Stimulated Bovine Endometrial Epithelial Cells Through Wnt/β-Catenin and PI3K/AKT Pathways. Front Vet Sci 2021; 8:637707. [PMID: 34307514 PMCID: PMC8299055 DOI: 10.3389/fvets.2021.637707] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/19/2021] [Indexed: 11/17/2022] Open
Abstract
Meloxicam is a non-steroidal anti-inflammatory drug and has been used to relieve pain and control inflammation in cows with metritis and endometritis. Meloxicam has been found to be effective in inhibiting tissue or cell growth when it is used as an anti-inflammatory therapy. However, the influence of meloxicam on bovine endometrial regeneration has not been reported. This study was to research the effect of meloxicam (0.5 and 5 μM) on the proliferation of primary bovine endometrial epithelial cells (BEECs) stimulated by Escherichia coli lipopolysaccharide. The cell viability, cell cycle, and cell proliferation were evaluated by Cell Counting Kit-8, flow cytometry, and cell scratch test, respectively. The mRNA transcriptions of prostaglandin-endoperoxide synthase 1 (PTGS1) and PTGS2, Toll-like receptor 4, and proliferation factors were detected using quantitative reverse-transcription polymerase chain reaction. The activations of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and Wnt/β-catenin pathways were determined using western blot and immunofluorescence. As a result, co-treatment of meloxicam and lipopolysaccharide inhibited (P < 0.05) the cell cycle progression and reduced (P < 0.05) the cell healing rate and the mRNA level of proliferation factors as compared with the cells treated with lipopolysaccharide alone. Meloxicam decreased (P < 0.05) the lipopolysaccharide-induced PTGS2 gene expression. Neither lipopolysaccharide nor meloxicam changed PTGS1 mRNA abundance (P > 0.05). Meloxicam inhibited (P < 0.05) the lipopolysaccharide-activated Wnt/β-catenin pathway by reducing (P < 0.05) the protein levels of β-catenin, c-Myc, cyclin D1, and glycogen synthase kinase-3β and prevented the lipopolysaccharide-induced β-catenin from entering the nucleus. Meloxicam suppressed (P < 0.05) the phosphorylation of PI3K and AKT. In conclusion, meloxicam alone did not influence the cell cycle progression or the cell proliferation in BEEC but caused cell cycle arrest and inhibited cell proliferation in lipopolysaccharide-stimulated BEEC. This inhibitory effect of meloxicam was probably mediated by Wnt/β-catenin and PI3K/AKT pathways.
Collapse
Affiliation(s)
- Luying Cui
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| | - Yang Qu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| | - Hele Cai
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| | - Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| | - Jun Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| | - Chen Qian
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, China
| |
Collapse
|
9
|
Anti-inflammatory and cytoprotective potentials of Meloxicam solid dispersions prepared by different techniques on lipopolysaccharide-stimulated RAW 264.7 macrophages. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
10
|
Gowifel AMH, Khalil MG, Nada SA, Kenawy SA, Ahmed KA, Salama MM, Safar MM. Combination of pomegranate extract and curcumin ameliorates thioacetamide-induced liver fibrosis in rats: impact on TGF-β/Smad3 and NF-κB signaling pathways. Toxicol Mech Methods 2020; 30:620-633. [PMID: 32718261 DOI: 10.1080/15376516.2020.1801926] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protection against liver injury and its consequences is considered an essential issue to minimize the number of annual deaths caused by liver diseases. The present study was designed to evaluate the potential role of pomegranate extract (PE) and/or curcumin in the regression of thioacetamide (TAA)-induced liver fibrosis, focusing on their modulatory effects on Nrf2/HO-1, NF-κB, and TGF-β/Smad3 signaling pathways. Liver fibrosis was induced in male Wistar rats by intraperitoneal injection of TAA (100 mg/kg) three times a week, for 8 weeks. To assess the protective effects of PE and/or curcumin against TAA-induced liver fibrosis, rats were treated on a daily basis with oral doses of PE (200 mg/kg) and/or curcumin (200 mg/kg) for 8 weeks. The results indicated that PE and/or curcumin attenuated TAA-induced liver fibrogenesis, as evidenced by a significant improvement in the liver function tests (AST, ALT, ALP, and albumin), oxidative stress biomarkers (MDA, SOD, and GSH), and inflammatory biomarkers (NF-ĸB, TNF-α, IL-1β, iNOS, TGF-β, and MPO), compared to TAA group. Moreover, treatment with PE and/or curcumin exerted a significant upregulation of Nrf2/HO-1 gene expressions along with significant downregulation of NF-ĸB, TGF-β, and phospho-Smad3 protein expressions, as well as α-SMA and collagen-1 gene expressions. The histopathological examination has corroborated these findings. In conclusion, hepatoprotective activities of PE and/or curcumin could be linked to their abilities to modulate Nrf2/HO-1, NF-κB, and TGF-β/Smad3 signaling pathways. It is worth noting that the combination of PE and curcumin exerted superior hepatoprotective effects against TAA-induced liver fibrosis, as compared to monotherapy.
Collapse
Affiliation(s)
- Ayah M H Gowifel
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Modern University for Technology and Information, Cairo, Egypt
| | - Mona G Khalil
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Modern University for Technology and Information, Cairo, Egypt
| | - Somaia A Nada
- Pharmacology Department, National Research Centre, Giza, Egypt
| | - Sanaa A Kenawy
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Cairo University, Cairo, Egypt
| | - Kawkab A Ahmed
- Faculty of Veterinary Medicine, Pathology Department, Cairo University, Giza, Egypt
| | - Maha M Salama
- Faculty of Pharmacy, Department of Pharmacognosy, Cairo University, Cairo, Egypt.,Faculty of Pharmacy, Department of Pharmacognosy, The British University in Egypt, Cairo, Egypt
| | - Marwa M Safar
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Cairo University, Cairo, Egypt.,Faculty of Pharmacy, Pharmacology and Biochemistry Department, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
11
|
Ismail RS, El-Awady MS, Hassan MH. Pantoprazole abrogated cisplatin-induced nephrotoxicity in mice via suppression of inflammation, apoptosis, and oxidative stress. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1161-1171. [PMID: 31950223 DOI: 10.1007/s00210-020-01823-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 01/10/2020] [Indexed: 12/17/2022]
Abstract
The current study was designed to evaluate the potential abatement effect of pantoprazole against cisplatin-induced nephrotoxicity and establishing the possible protective mechanisms. Thirty-two male mice were allocated for treatment with saline, single dose of cisplatin (10 mg/kg/i.p), pantoprazole (30 mg/kg/once daily) for 5 days or combination of pantoprazole and cisplatin for 5 days. Urine, blood, and both kidneys were collected for further evaluations. Pantoprazole significantly countermand cisplatin-induced disfigurement of renal histology, kidney weight to body weight ratio, serum levels of creatinine and urea, and microalbuminuria. Furthermore, pantoprazole mostly normalized cisplatin-induced distortion of renal levels of inflammatory cytokines (tumor necrosis factor-alpha, interleukin-6, interleukin-10) and renal content of apoptosis regulating protein expressions (Bax, Bcl2, and active caspase 3). In addition, pantoprazole significantly subsided cisplatin-induced distortion of renal lipid peroxidation marker (MDA), renal superoxide dismutase, and catalase activities and renal reduced glutathione content. This study provides an evidence for the protective utility of short-term pantoprazole against cisplatin-induced nephrotoxicity in mice. The protective mechanism of pantoprazole could be through diminution of cisplatin-induced inflammation, oxidative stress, and their subsequent apoptotic renal cell death via abatement of apoptosis regulating protein expressions (Bax, Bcl2, and active caspase3).
Collapse
Affiliation(s)
- Raed S Ismail
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azahr University, Cairo, Egypt
| | - Mohammed S El-Awady
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawara, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Memy H Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azahr University, Cairo, Egypt.
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawara, Saudi Arabia.
| |
Collapse
|
12
|
Nagy EE, Frigy A, Szász JA, Horváth E. Neuroinflammation and microglia/macrophage phenotype modulate the molecular background of post-stroke depression: A literature review. Exp Ther Med 2020; 20:2510-2523. [PMID: 32765743 PMCID: PMC7401670 DOI: 10.3892/etm.2020.8933] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence hints to the central role of neuroinflammation in the development of post-stroke depression. Danger signals released in the acute phase of ischemia trigger microglial activation, along with the infiltration of neutrophils and macrophages. The increased secretion of proinflammatory cytokines interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor α (TNFα) provokes neuronal degeneration and apoptosis, whereas IL-6, interferon γ (IFNγ), and TNFα induce aberrant tryptophane degradation with the accumulation of the end-product quinolinic acid in resident glial cells. This promotes glutamate excitotoxicity via hyperexcitation of N-methyl-D-aspartate receptors and antagonizes 5-hydroxy-tryptamine, reducing synaptic plasticity and neuronal survival, thus favoring depression. In the post-stroke period, CX3CL1 and the CD200-CD200R interaction mediates the activation of glial cells, whereas CCL-2 attracts infiltrating macrophages. CD206 positive cells grant the removal of excessive danger signals; the high number of regulatory T cells, IL-4, IL-10, transforming growth factor β (TGFβ), and intracellular signaling via cAMP response element-binding protein (CREB) support the M2 type differentiation. In favorable conditions, these cells may exert efficient clearance, mediate tissue repair, and might be essential players in the downregulation of molecular pathways that promote post-stroke depression.
Collapse
Affiliation(s)
- Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Attila Frigy
- Department of Internal Medicine IV, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - József Attila Szász
- Neurology Clinic II, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540136 Targu Mures, Romania
| | - Emőke Horváth
- Department of Pathology, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| |
Collapse
|
13
|
da Silva JCG, Dallegrave E, Rodrigues GZP, Bigolin C, Neumann TMSDO, Schuster AC, Kayser JM, Staudt LBM, Moraes MF, Farias DG, Schiling G, Raasch JR, Perassolo MS, da Silva LB, Gehlen G, Betti AH. Repeated dose of meloxicam induces genotoxicity and histopathological changes in cardiac tissue of mice. Drug Chem Toxicol 2020; 45:822-833. [PMID: 32552192 DOI: 10.1080/01480545.2020.1778018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Meloxicam is the non-steroidal anti-inflammatory drug most used in small animals; however, studies on genotoxicity, oxidative stress, and histopathologic alterations in cardiac tissue are limited, especially at therapeutical doses used in these animals. This study evaluated the toxic effects caused by the treatment involving repeated low at higher doses of meloxicam in mice, by genotoxicity, oxidative stress, and histopathological parameters. Mice (CF1, male) received, by gavage, meloxicam at the therapeutic dose indicated for small animals (0.1 mg/kg) and at higher doses (0.5 and 1 mg/kg) for 28 days. Later, they were euthanized for blood and organ analysis. Oxidative stress was analyzed by the plasma ferric reduction capacity (FRAP) and catalase, and genotoxicity, by the comet assay and the micronucleus test. Heart, liver, lung, and kidney tissues were analyzed by the histology, and stomach and duodenum were analyzed with a magnifying glass. The relative weight of organs did not present significant alterations. However, congestion of duodenum vessels was observed at the three tested doses and caused hyperemia of stomach mucosa at 1 mg/kg. In the heart histology there was a reduction in the number of cardiomyocytes, accompanied by an increase in cell diameter (possible cell hypertrophy) dose-dependent. The highest tested dose of meloxicam also increased the DNA damage index, without alterations in the micronucleus test. Meloxicam did not affect the catalase activity but increased the FRAP (1 mg/kg). Meloxicam at the dose prescribed for small animals could potentially cause cardiac histopathologic alterations and genotoxic effects.
Collapse
Affiliation(s)
| | - Eliane Dallegrave
- Toxicology Department, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Cassiana Bigolin
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | | | - Andriéli Carolina Schuster
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Juliana Machado Kayser
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | | | - Melina Floriano Moraes
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Débora Graziela Farias
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Gabriela Schiling
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Juliana Raquel Raasch
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Magda Susana Perassolo
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Luciano Basso da Silva
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Günther Gehlen
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Andresa Heemann Betti
- Bioanalysis Department, Institute of Health Sciences, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| |
Collapse
|
14
|
Li J, Fan X, Deng J, Liang Y, Ma S, Lu Y, Zhang J, Shi T, Tan W, Wang Z. Design and synthesis of 1,3-benzothiazinone derivatives as potential anti-inflammatory agents. Bioorg Med Chem 2020; 28:115526. [PMID: 32354672 DOI: 10.1016/j.bmc.2020.115526] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 02/09/2023]
Abstract
A series of 1,3-benzothiazinone derivatives were designed and synthesized for pharmacological assessments. Among the synthesized 19 compounds, some compounds showed high activities on inhibiting LPS-induced nitrite oxide and TNF-α production, down-regulating COX-2 and increasing IL-10 production in RAW264.7 cells. All the compounds had no obvious cytotoxicity in in vitro assay. LD50 value of compound 25 was greater than 2000 mg/kg, which was safer than meloxicam. Compound 25 significantly inhibited phosphorylation of NF-κB and STAT3 in LPS-induced RAW264.7 cells. Inhibition of synthesized compounds on COX activity was weaker than meloxicam. Compound 25 displayed lower gastrointestinal toxicity than meloxicam. Besides, compound 25 decreased the swelling in carrageenan-induced paw edema models of inflammation and reduced PGE2 level significantly. In summary, 1,3-benzothiazinone derivatives are unique scaffolds with anti-inflammatory activity and low toxicity.
Collapse
Affiliation(s)
- Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiaohong Fan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jiedan Deng
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yan Liang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Shumeng Ma
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yingmei Lu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jian Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Tao Shi
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Zhen Wang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
15
|
Rehbein S, Manchi G, Gruber AD, Kohn B. Successful Treatment of Pneumothorax in a Dog With Sterile Pleural Fibrosis Caused by Chylothorax. Front Vet Sci 2019; 6:278. [PMID: 31508433 PMCID: PMC6713890 DOI: 10.3389/fvets.2019.00278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/07/2019] [Indexed: 01/30/2023] Open
Abstract
A 2-year-old, 12 kg, intact male crossbreed dog was presented with respiratory distress, exercise intolerance, and gagging. Plain thoracic radiographs revealed severe pleural effusion. Although bilateral needle thoracocentesis and chest tube placement were performed, no re-expansion of the lung lobes occurred. Pleural effusion was of chylous quality and led to lung entrapment. Computer tomography revealed a highly atrophic and atelectatic right middle lung lobe. The remaining lung lobes were only expanded to ~40%. Visceral pleura and pericardium showed a heterogeneous thickening consistent with pleural fibrosis. Partial pericardiectomy with resection of the middle lung lobe through a right lateral thoracotomy was performed. Ligation of the thoracic duct and ablation of the cisterna chyli was achieved through a single paracostal approach. Histopathology revealed chronic-active proliferative beginning granulomatous pleuritis, fibrotic pericarditis, and partial coagulative necrosis with incomplete granulomatous sequestration in the resected middle lung lobe. Chylothorax resolved after surgical intervention. Active pleural effusion resolved, and lung entrapment changed to trapped lung disease. The remaining lung lobes re-expanded to ~80% over the following 6 days. The dog was discharged 10 days later. Mild to moderate pleural effusion of non-chylic quality was present during the following 4 months. Meloxicam was administered for 4 months because of its anti-fibrotic and anti-inflammatory properties. Fifteen months later, thoracic radiographs revealed full radiologic expansion of the lungs with persistent mild pleural fibrosis. To the authors' knowledge, this is the first case report of pneumothorax due pleural fibrosis caused by chylothorax in a dog with an excellent clinical outcome.
Collapse
Affiliation(s)
- Sina Rehbein
- Clinic for Small Animals, Freie Universität, Berlin, Germany
| | - George Manchi
- Clinic for Small Animals, Freie Universität, Berlin, Germany
| | - Achim D. Gruber
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität, Berlin, Germany
| | - Barbara Kohn
- Clinic for Small Animals, Freie Universität, Berlin, Germany
| |
Collapse
|
16
|
Marinho TDS, Ornellas F, Barbosa-da-Silva S, Mandarim-de-Lacerda CA, Aguila MB. Beneficial effects of intermittent fasting on steatosis and inflammation of the liver in mice fed a high-fat or a high-fructose diet. Nutrition 2019; 65:103-112. [PMID: 31079017 DOI: 10.1016/j.nut.2019.02.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 01/29/2019] [Accepted: 02/23/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Intermittent fasting (IF) is a nutritional intervention with significant metabolic effects on the liver that are not yet fully understood. The aim of this study was to investigate the effects of IF on body mass, lipid profile, glucose metabolism, liver lipogenesis, β-oxidation, and inflammation. METHODS We used cellular and molecular techniques to investigate the effects of IF on 3-mo-old male C57 BL/6 mice that were fed control (10% kcal fat), high-fat (HF; 50% kcal fat), or high-fructose (HFr; 50% kcal fructose) diets for 8 wk. Half of the animals were submitted to IF (1 d fed, 1 d fast) for an additional 4 wk. RESULTS Although food intake on the fed day did not differ between the groups, mice in the HF and HFr groups showed diminished body mass, total cholesterol, and triacylglycerol levels. Also, plasma adiponectin increased in the HFr group and leptin decreased in the HF mice. Oral glucose tolerance test and insulin were ameliorated by IF, regardless of the diet consumed (HF or HFr), and decreased hepatic lipogenesis and increased β-oxidation markers, resulting in a reduction of the hepatic steatosis and inflammation. CONCLUSIONS There were beneficial effects of IF even with the continuity of the obesogenic diet and proinflammatory diet in mice. It is recommended that based on the beneficial effects of IF on glucose and liver metabolism and inflammation that IF be a coadjutant factor in the treatment of hepatic metabolic issues and steatosis.
Collapse
Affiliation(s)
- Thatiany de Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ornellas
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sandra Barbosa-da-Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
Mortezaee K, Majidpoor J, Daneshi E, Abouzaripour M, Abdi M. Post-treatment of melatonin with CCl 4 better reduces fibrogenic and oxidative changes in liver than melatonin co-treatment. J Cell Biochem 2018; 119:1716-1725. [PMID: 28782839 DOI: 10.1002/jcb.26331] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/04/2017] [Indexed: 08/28/2024]
Abstract
Therapeutic effects of melatonin (MEL) in targeting CCl4 -induced liver fibrosis has been widely known, but there is no study comparing oxidative and fibrogenic changes in co- and post-treatment of MEL with CCl4 , which was further aimed in this experiment. Male SD rats were injected with CCl4 (1 mL/kg/i.p./daily) dissolved 1:1 in olive oil for 1 month. Some animals received MEL (20 mg/kg/i.p./daily) diluted in 1 mL PBS in combination with CCl4 (co-treatment), and some rats were treated with MEL, beginning with injection of the last dose of CCl4 for one month (post-treatment). The groups were control, CCl4 , CCl4 -co vehicle, CCl4 -post vehicle, post-CCl4 , MEL co-treatment, and MEL post-treatment. MEL post-treatment group showed significantly lower lipid deposition, serum malondialdehyde (MDA), serum alanine aminotransferase (ALT), and liver hydroxyproline. This group also had low expressions of Bax and transforming growth factor-β1 (TGF-β1). MEL post-treatment group revealed higher sera levels of albumin, superoxide dismutase (SOD) and glutathione peroxidase (GPx). Expression levels of metalloproteinase-13 (MMP-13) and Bcl2 was also higher in this group (P ≤ 0.05 vs co-treatment). Results of the present study indicated that MEL post-treatment is more powerful in reduction of CCl4 -induced liver fibrosis through reduction of oxidative stress and maintenance of matrix balance.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Daneshi
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Morteza Abouzaripour
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mahdad Abdi
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|